Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharmacol ; 95(2): 169-182, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30409791

RESUMEN

Organic cation transporters OCT1 (SLC22A1) and OCT2 (SLC22A2) are critically involved in absorption and excretion of diverse cationic drugs. Because drug-drug interactions at these transporters may induce adverse drug effects in patients, in vitro testing during drug development for interaction with the human transporters is mandatory. Recent data performed with rat OCT1 (rOCT1) suggest that currently performed in vitro tests assuming one polyspecific binding site are insufficient. Here we measured the binding and transport of model substrate 1-methyl-4-phenylpyridinium+ (MPP+) by cell-free-expressed fusion proteins of rOCT1 and rOCT1 mutants with green fluorescent protein that had been reconstituted into nanodiscs or proteoliposomes. The nanodiscs were formed with major scaffold protein (MSP) and different phospholipids, whereas the proteoliposomes were formed with a mixture of cholesterol, phosphatidylserine, and phosphatidylcholine. In nanodiscs formed with 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine or cholesterol, phosphatidylserine, and phosphatidylcholine, two low-affinity MPP+ binding sites and one high-affinity MPP+ binding site per transporter monomer were determined. Mutagenesis revealed that tryptophan 218 and aspartate 475 in neighboring positions in the modeled outward-open cleft contribute to one low-affinity binding site, whereas arginine 440 located distantly in the cleft is critical for MPP+ binding to another low-affinity site. Comparing MPP+ binding with MPP+ transport suggests that the low-affinity sites are involved in MPP+ transport, whereas high-affinity MPP+ binding influences transport allosterically. The data will be helpful in the interpretation of future crystal structures and provides a rationale for future in vitro testing that is more sophisticated and reliable, leading to the generation of pharmacophore models with high predictive power.


Asunto(s)
1-Metil-4-fenilpiridinio/metabolismo , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Animales , Sitios de Unión , Proteínas Fluorescentes Verdes/metabolismo , Mutagénesis/fisiología , Fosfolípidos/metabolismo , Proteolípidos/metabolismo , Ratas
2.
Mol Pharmacol ; 93(4): 402-415, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29339398

RESUMEN

The effects of mutations in the modeled outward-open cleft of rat organic cation transporter 1 (rOCT1) on affinities of substrates and inhibitors were investigated. Human embryonic kidney 293 cells were stably transfected with rOCT1 or rOCT1 mutants, and uptake of the substrates 1-methyl-4-phenylpyridinium+ (MPP+) and tetraethylammonium+ (TEA+) or inhibition of MPP+ uptake by the nontransported inhibitors tetrabutylammonium+ (TBuA+), tetrapentylammonium+ (TPeA+), and corticosterone was measured. Uptake measurements were performed on confluent cell layers using a 2-minute incubation or in dissociated cells using incubation times of 1, 5, or 10 seconds. With both methods, different apparent Michaelis-Menten constant (Km) values, different IC50 values, and varying effects of mutations were determined. In addition, varying IC50 values for the inhibition of MPP+ uptake and varying effects of mutations were obtained when different MPP+ concentrations far below the apparent Km value were used for uptake measurements. Eleven mutations were investigated by measuring initial uptake in dissociated cells and employing 0.1 µM MPP+ for uptake during inhibition experiments. Altered affinities for substrates and/or inhibitors were observed when Phe160, Trp218, Arg440, Leu447, and Asp475 were mutated. The mutations resulted in changes of apparent Km values for TEA+ and/or MPP+ Mutation of Trp218 and Asp475 led to altered IC50 values for TBuA+, TPeA+, and corticosterone, whereas the mutation of Phe160 and Leu447 changed the IC50 values for two inhibitors. Thereby amino acids in the outward-facing conformation of rOCT1 could be identified that interact with structurally different inhibitors and probably also with different substrates.


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/antagonistas & inhibidores , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Mutagénesis/efectos de los fármacos , 1-Metil-4-fenilpiridinio/metabolismo , 1-Metil-4-fenilpiridinio/farmacología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Células HEK293 , Humanos , Mutagénesis/fisiología , Compuestos de Amonio Cuaternario/metabolismo , Compuestos de Amonio Cuaternario/farmacología , Ratas , Especificidad por Sustrato/efectos de los fármacos , Especificidad por Sustrato/fisiología , Xenopus laevis
3.
Mol Pharmacol ; 89(1): 118-32, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26464324

RESUMEN

Cellular uptake adapts rapidly to physiologic demands by changing transporter abundance in the plasma membrane. The human gene RSC1A1 codes for a 67-kDa protein named RS1 that has been shown to induce downregulation of the sodium-D-glucose cotransporter 1 (SGLT1) and of the concentrative nucleoside transporter 1 (CNT1) in the plasma membrane by blocking exocytosis at the Golgi. Injecting RS1 fragments into Xenopus laevis oocytes expressing SGLT1 or CNT1 and measuring the expressed uptake of α-methylglucoside or uridine 1 hour later, we identified a RS1 domain (RS1-Reg) containing multiple predicted phosphorylation sites that is responsible for this post-translational downregulation of SGLT1 and CNT1. Dependent on phosphorylation, RS1-Reg blocks the release of SGLT1-containing vesicles from the Golgi in a glucose-dependent manner or glucose-independent release of CNT1-containing vesicles. We showed that upregulation of SGLT1 in the small intestine after glucose ingestion is promoted by glucose-dependent disinhibition of the RS1-Reg-blocked exocytotic pathway of SGLT1 between meals. Mimicking phosphorylation of RS1-Reg, we obtained a RS1-Reg variant that downregulates SGLT1 in the brush-border membrane at high luminal glucose concentration. Because RS1 mediates short-term regulation of various transporters, we propose that the RS1-Reg-navigated transporter release from Golgi represents a basic regulatory mechanism of general importance, which implies the existence of receptor proteins that recognize different phosphorylated forms of RS1-Reg and of complex transporter-specific sorting in the trans-Golgi. RS1-Reg-derived peptides that downregulate SGLT1 at high intracellular glucose concentrations may be used for downregulation of glucose absorption in small intestine, which has been proposed as strategy for treatment of type 2 diabetes.


Asunto(s)
Exocitosis/fisiología , Glucosa/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Transducción de Señal/fisiología , Transportador 1 de Sodio-Glucosa/metabolismo , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación/fisiología , Transportador 1 de Sodio-Glucosa/antagonistas & inhibidores , Xenopus laevis
4.
Mol Pharmacol ; 90(5): 508-521, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27555600

RESUMEN

Na+-d-glucose cotransporter 1 (SGLT1) is rate-limiting for glucose absorption in the small intestine. Shortly after intake of glucose-rich food, SGLT1 abundance in the luminal membrane of the small intestine is increased. This upregulation occurs via glucose-induced acceleration of the release of SGLT1-containing vesicles from the trans-Golgi network (TGN), which is regulated by a domain of protein RS1 (RSC1A1) named RS1-Reg. Dependent on phosphorylation, RS1-Reg blocks release of vesicles containing SGLT1 or concentrative nucleoside transporter 1. The hypothesis has been raised that RS1-Reg binds to different receptor proteins at the TGN, which trigger release of vesicles with different transporters. To identify the presumed receptor proteins, two-hybrid screening was performed. Interaction with ornithine decarboxylase 1 (ODC1), the rate-limiting enzyme of polyamine synthesis, was observed and verified by immunoprecipitation. Binding of RS1-Reg mutants to ODC1 was characterized using surface plasmon resonance. Inhibition of ODC1 activity by RS1-Reg mutants and the ODC1 inhibitor difluoromethylornithine (DFMO) was measured in the absence and presence of glucose. In addition, short-term effects of DFMO, RS1-Reg mutants, the ODC1 product putrescine, and/or glucose on SGLT1 expressed in oocytes of Xenopus laevis were investigated. High-affinity binding of RS1-Reg to ODC1 was demonstrated, and evidence for a glucose binding site in ODC1 was provided. Binding of RS1-Reg to ODC1 inhibits the enzymatic activity at low intracellular glucose, which is blunted at high intracellular glucose. The data suggest that generation of putrescine by ODC1 at the TGN stimulates release of SGLT1-containing vesicles. This indicates a biomedically important role of ODC1 in regulation of glucose homeostasis.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Exocitosis/efectos de los fármacos , Glucosa/farmacología , Proteínas de Transporte de Monosacáridos/metabolismo , Ornitina Descarboxilasa/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Eflornitina/farmacología , Electroforesis en Gel de Poliacrilamida , Células HEK293 , Humanos , Inmunoprecipitación , Espacio Intracelular/metabolismo , Cinética , Metilglucósidos/farmacología , Modelos Biológicos , Proteínas de Transporte de Monosacáridos/química , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Florizina/farmacología , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Proteínas Recombinantes/metabolismo , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Resonancia por Plasmón de Superficie , Xenopus laevis
5.
Clin Immunol ; 158(1): 47-58, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25796192

RESUMEN

In multiple sclerosis (MS) lymphoid follicle-like aggregates have been reported in the meninges of patients. Here we investigated the functional relevance of B cell infiltration into the central nervous system (CNS) in MP4-induced experimental autoimmune encephalomyelitis (EAE), a B cell-dependent mouse model of MS. In chronic EAE, B cell aggregates were characterized by the presence of CXCL13(+) and germinal center CD10(+) B cells. Germline transcripts were expressed in the CNS and particularly related to TH17-associated isotypes. We also observed B cells with restricted VH gene usage that differed from clones found in the spleen. Finally, we detected CNS-restricted spreading of the antigen-specific B cell response towards a myelin and a neuronal autoantigen. These data imply the development of autonomous B cell-mediated autoimmunity in the CNS in EAE - a concept that might also apply to MS itself.


Asunto(s)
Autoantígenos/inmunología , Linfocitos B/inmunología , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Vaina de Mielina/inmunología , ARN Mensajero/metabolismo , Animales , Cerebelo/inmunología , Cerebelo/metabolismo , Quimiocina CXCL13/inmunología , Modelos Animales de Enfermedad , Ratones , Proteína Básica de Mielina/inmunología , Proteína Proteolipídica de la Mielina/inmunología , Neprilisina/inmunología , Bazo/inmunología , Bazo/metabolismo
6.
J Biol Chem ; 287(37): 31561-73, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22810231

RESUMEN

Organic cation transporters are membrane potential-dependent facilitative diffusion systems. Functional studies, extensive mutagenesis, and homology modeling indicate the following mechanism. A transporter conformation with a large outward-open cleft binds extracellular substrate, passes a state in which the substrate is occluded, turns to a conformation with an inward-open cleft, releases substrate, and subsequently turns back to the outward-open state. In the rat organic cation transporter (rOct1), voltage- and ligand-dependent movements of fluorescence-labeled cysteines were measured by voltage clamp fluorometry. For fluorescence detection, cysteine residues were introduced in extracellular parts of cleft-forming transmembrane α-helices (TMHs) 5, 8, and 11. Following expression of the mutants in Xenopus laevis oocytes, cysteines were labeled with tetramethylrhodamine-6-maleimide, and voltage-dependent conformational changes were monitored by voltage clamp fluorometry. One cysteine was introduced in the central domain of TMH 11 replacing glycine 478. This domain contains two amino acids that are involved in substrate binding and two glycine residues (Gly-477 and Gly-478) allowing for helix bending. Cys-478 could be modified with the transported substrate analog [2-(trimethylammonium)-ethyl]methanethiosulfonate but was inaccessible to tetramethylrhodamine-6-maleimide. Voltage-dependent movements at the indicator positions of TMHs 5, 8, and 11 were altered by substrate applications indicating large conformational changes during transport. The G478C exchange decreased transporter turnover and blocked voltage-dependent movements of TMHs 5 and 11. [2-(Trimethylammonium)-ethyl]methanethiosulfonate modification of Cys-478 blocked substrate binding, transport activity, and movement of TMH 8. The data suggest that Gly-478 is located within a mechanistically important hinge domain of TMH 11 in which substrate binding induces transport-related structural changes.


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Activación del Canal Iónico/fisiología , Animales , Sitios de Unión , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/química , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Células HEK293 , Humanos , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Especificidad por Sustrato/fisiología , Xenopus laevis
7.
Mol Pharm ; 10(8): 3045-56, 2013 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-23763587

RESUMEN

Polyspecific organic cation transporter Oct2 from rat (gene Slc22A2) has been previously shown to transport Cs(+). Here we report that human OCT2 (hOCT2) is able to transport Cd(2+) showing substrate saturation with a Michaelis-Menten constant (Km) of 54 ± 5.8 µM. Uptake of Cd(2+) by hOCT2 was inhibited by typical hOCT2 ligands (unlabeled substrates and inhibitors), and the rate of uptake was decreased by a point mutation in a substrate binding domain of hOCT2. Incubation of hOCT2 overexpressing human embryonic kidney 293 cells (HEK-hOCT2-C) or rat renal proximal tubule cells expressing rOct2 (NRK-52E-C) with Cd(2+) resulted in an increased level of apoptosis that was reduced by OCT2 inhibitory ligand cimetidine(+). HEK-hOCT2-C exhibited different functional properties when they were confluent or had been dissociated by removal of Ca(2+) and Mg(2+). Only confluent HEK-hOCT2-C transported Cd(2+), and confluent and dissociated cells exhibited different potencies for inhibition of uptake of 1-methyl-4-phenylpyridinium(+) (MPP(+)) by Cd(2+), MPP(+), tetraethylammonium(+), cimetidine(+), and corticosterone. In confluent HEK-hOCT2-C, largely different inhibitor potencies were obtained upon comparison of inhibition of Cd(2+) uptake, 4-[4-(dimethylamino)styryl]-N-methylpyridinium(+) (ASP(+)) uptake, and MPP(+) uptake using substrate concentrations far below the respective Km values. Employing a point mutation in the previously identified substrate binding site of rat Oct1 produced evidence that short distance allosteric effects between binding sites for substrates and inhibitors are involved in substrate-dependent inhibitor potency. Substrate-dependent inhibitor affinity is probably a common property of OCTs. To predict interactions between drugs that are transported by OCTs and inhibitory drugs, it is necessary to employ the specific transported drug rather than a model substrate for in vitro measurements.


Asunto(s)
Cadmio/metabolismo , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Proteínas de Transporte de Catión Orgánico/metabolismo , Animales , Apoptosis/efectos de los fármacos , Cadmio/farmacología , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Línea Celular , Cimetidina/farmacología , Corticosterona/farmacología , Humanos , Túbulos Renales Proximales/metabolismo , Transportador 2 de Cátion Orgánico , Ratas , Tetraetilamonio/farmacología
8.
J Biol Chem ; 286(43): 37874-86, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21896487

RESUMEN

Polyspecific organic anion transporters (OATs) and organic cation transporters (OCTs) of the SLC22 transporter family play a pivotal role in absorption, distribution, and excretion of drugs. Polymorphisms in these transporters influence therapeutic effects. On the basis of functional characterizations, homology modeling, and mutagenesis, hypotheses for how OCTs bind and translocate structurally different cations were raised, assuming functionally competent monomers. However, homo-oligomerization has been described for OATs and OCTs. In the present study, evidence is provided that the large extracellular loops (EL) of rat Oct1 (rOct1) and rat Oat1 (rOat1) mediate homo- but not hetero-oligomerization. Replacement of the cysteine residues in the EL of rOct1 by serine residues (rOct1(6ΔC-l)) or breaking disulfide bonds with dithiothreitol prevented oligomerization. rOct1 chimera containing the EL of rOat1 (rOct1(rOat1-l)) showed oligomerization but reduced transporter amount in the plasma membrane. For rOct1(6ΔC-l) and rOct1(rOat1-l), similar K(m) values for 1-methyl-4-phenylpyridinium(+) (MPP(+)) and tetraethylammonium(+) (TEA(+)) were obtained that were higher compared with rOct1 wild type. The increased K(m) of rOct1(rOat1-l) indicates an allosteric effect of EL on the cation binding region. The similar substrate affinity of the oligomerizing and non-oligomerizing loop mutants suggests that oligomerization does not influence transport function. Independent transport function of rOct1 monomers was also demonstrated by showing that K(m) values for MPP(+) and TEA(+) were not changed after treatment with dithiothreitol and that a tandem protein with two rOct1 monomers showed about 50% activity with unchanged K(m) values for MPP(+) and TEA(+) when one monomer was blocked. The data help to understand how OCTs work and how mutations in patients may affect their functions.


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Multimerización de Proteína/fisiología , Animales , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/química , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Ditiotreitol/química , Ditiotreitol/farmacología , Células HEK293 , Humanos , Transporte Iónico/efectos de los fármacos , Transporte Iónico/fisiología , Mutación , Proteína 1 de Transporte de Anión Orgánico/química , Proteína 1 de Transporte de Anión Orgánico/genética , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Multimerización de Proteína/efectos de los fármacos , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Xenopus laevis
9.
Traffic ; 10(11): 1599-618, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19765263

RESUMEN

The gene product of RSC1A1, RS1, participates in the regulation of the Na(+)-D-glucose cotransporter SGLT1. RS1 inhibits release of SGLT1 from the trans Golgi network. In subconfluent LLC-PK(1) cells, RS1 migrates into the nucleus and modulates transcription of SGLT1, whereas most confluent cells do not contain RS1 in the nuclei. We showed that confluence-dependent nuclear location of RS1 is because of different phases of the cell cycle and identified a RS1 nuclear shuttling domain (RNS) with an associated protein kinase C (PKC) phosphorylation site (RNS-PKC) that mediates cell cycle-dependent nuclear location. RNS-PKC contains a novel non-conventional nuclear localization signal interacting with importin beta1, a nuclear export signal mediating export via protein CRM1 and a Ca(2+)-dependent calmodulin binding site. PKC and calmodulin compete for binding to RNS-PKC. Mutagenesis experiments and analyses of the phosphorylation status suggest the following sequences of events. Subconfluent cells without and with synchronization to the G2/M phase contain non-phosphorylated RNS-PKC that mediates nuclear import of RS1 but not its export. During confluence or synchronization of subconfluent cells to the G2/M phase, phosphorylation of RNS-PKC mediates rapid nuclear export of RS1.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Monosacáridos/química , Proteínas de Transporte de Monosacáridos/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo , Transporte Activo de Núcleo Celular , Animales , Ciclo Celular , Medio de Cultivo Libre de Suero , Vectores Genéticos , Glucosa/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Carioferinas/metabolismo , Células LLC-PK1 , Proteínas de Transporte de Monosacáridos/genética , Señales de Localización Nuclear/metabolismo , Fosforilación , Proteína Quinasa C/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas/fisiología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Sodio/metabolismo , Porcinos , Factores de Tiempo , Transfección , Red trans-Golgi/metabolismo
10.
Mol Pharmacol ; 76(2): 275-89, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19435783

RESUMEN

We have shown previously that Leu447 and Gln448 in the transmembrane helix (TMH) 10 of rat organic cation transporter rOCT1 are critical for inhibition of cation uptake by corticosterone. Here, we tested whether the affinity of corticosterone is different when applied from the extracellular or intracellular side. The affinity of corticosterone was determined by measuring the inhibition of currents induced by tetraethylammonium(+) (TEA(+)) in Xenopus laevis oocytes expressing rOCT1. Either corticosterone and TEA(+) were added to the bath simultaneously or the oocytes were preincubated with corticosterone, washed, and TEA(+)-induced currents were determined subsequently. In mutant L447Y, K(i) values for extracellular and intracellular corticosterone were decreased, whereas in mutant Q448E, only the K(i) for intracellular corticosterone was changed. Modeling of the interaction of corticosterone with rOCT1 in the inward- or outward-facing conformation predicted direct binding to Leu447, Phe160 (TMH2), Trp218 (TMH4), Arg440 (TMH10), and Asp475 (TM11) from both sides. In mutant F160A, affinities for extracellular and intracellular corticosterone were increased, whereas maximal inhibition was reduced in W218F and R440K. In stably transfected epithelial cells, the affinities for inhibition of 1-methyl-4-phenyl-pyridinium(+) (MPP(+)) uptake by extracellular and intracellular corticosterone were decreased when Asp475 was replaced by glutamate. In mutants F160A, W218Y, R440K, and L447F, the affinities for MPP(+) uptake were changed, and in mutant D475E, the affinity for TEA(+) uptake was changed. The data suggest that Phe160, Trp218, Arg440, Leu447, and Asp475 are located within an innermost cavity of the binding cleft that is alternatingly exposed to the extracellular or intracellular side during substrate transport.


Asunto(s)
Aminoácidos/metabolismo , Corticosterona/metabolismo , Transportador 1 de Catión Orgánico/metabolismo , Alanina/metabolismo , Secuencia de Aminoácidos/genética , Sustitución de Aminoácidos , Aminoácidos/genética , Animales , Corticosterona/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Concentración 50 Inhibidora , Modelos Moleculares , Datos de Secuencia Molecular , Oocitos , Transportador 1 de Catión Orgánico/genética , Técnicas de Placa-Clamp , Fenilalanina/metabolismo , Mutación Puntual , Estructura Secundaria de Proteína , Ratas , Homología de Secuencia de Aminoácido , Especificidad por Sustrato/fisiología , Tirosina/metabolismo , Xenopus laevis
11.
J Pharmacol Exp Ther ; 329(1): 252-61, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19141712

RESUMEN

Nucleoside reverse transcriptase inhibitors (NRTIs) need to enter cells to act against the HIV-1. Human organic cation transporters (hOCT1-3) are expressed and active in CD4+ T cells, the main target of HIV-1, and have been associated with antiviral uptake in different tissues. In this study, we examined whether NRTIs interact and are substrates of hOCT in cells stably expressing these transporters. Using [(3)H]N-methyl-4-phenylpyridinium, we found a high-affinity interaction among abacavir [[(1S,4R)-4-[2-amino-6-(cyclopropylamino)purin-9-yl]-cyclopent-2-enyl]methanol sulfate] (ABC); <0.08 nM], azidothymidine [3'-azido-3'-deoxythymidine (AZT); <0.4 nM], tenofovir disoproxil fumarate (<1.0 nM), and emtricitabine (<2.5 nM) and hOCTs. Using a wide range of concentrations of lamivudine [(-)-beta-L-2',3'-dideoxy-3'-thiacyitidine (3TC)], we determined two different binding sites for hOCTs: a high-affinity site (K(d1) = 12.3-15.4 pM) and a low-affinity site (K(d2) = 1.9-3.4 mM). Measuring direct uptake of [(3)H]3TC and inhibition with hOCT substrates, we identified 3TC as a novel substrate for hOCT1, 2, and 3, with hOCT1 as the most efficient transporter (K(m) = 1.25 +/- 0.1 mM; V(max) = 10.40 +/- 0.32 nmol/mg protein/min; V(max)/K(m) = 8.32 +/- 0.40 microl/mg protein/min). In drug-drug interaction experiments, we analyzed cis-inhibition of [(3)H]3TC uptake by ABC and AZT and found that 40 to 50% was inhibited at low concentrations of the drugs (K(i) = 22-500 pM). These data reveal that NRTIs experience a high-affinity interaction with hOCTs, suggesting a putative role for these drugs as modulators of hOCT activity. Finally, 3TC is a novel substrate for hOCTs and the inhibition of its uptake at low concentrations of ABC and AZT could have implications for the pharmacokinetics of 3TC.


Asunto(s)
Fármacos Anti-VIH/metabolismo , Lamivudine/metabolismo , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Inhibidores de la Transcriptasa Inversa/metabolismo , Animales , Sitios de Unión/efectos de los fármacos , Transporte Biológico Activo/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , Didesoxinucleósidos/metabolismo , Interacciones Farmacológicas , Humanos , Cinética , Transfección , Zidovudina/metabolismo
12.
Mol Pharmacol ; 73(1): 50-61, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17940192

RESUMEN

Voltage-clamp fluorometry was performed with a cysteine-deprived mutant of rat organic cation transporter 1 (rOCT1) in which Phe483 in transmembrane alpha-helix (TMH) 11 close to the extracellular surface was replaced by cysteine and labeled with tetramethylrhodamine-6-maleimide. Potential-dependent fluorescence changes were observed that were sensitive to presence of substrates choline, tetraethylammonium (TEA), and 1-methyl-4-phenylpyridinium (MPP) and of the nontransported inhibitor tetrabutylammonium (TBuA). Using potential-dependent fluorescence changes as readout, one high-affinity binding site per substrate and two high-affinity binding sites for TBuA were identified in addition to the previously described single interaction sites. In a structure model of rOCT1 with an inward open cleft that was derived from a known crystal structure of lacY permease, Phe483 is close to Trp147 in TMH 2. In contrast, in a model with an outward open cleft these amino acids are far apart. After replacement of Phe483 or Trp147 by cysteine or serine, high-affinity binding of TBuA leads to inhibition of MPP or TEA uptake, whereas it has no effect on cation uptake by wild-type rOCT1. Coexisting high-affinity cation binding sites in organic cation transporters may collect low concentration xenobiotics and drugs; however, translocation including transitions between outward- and inward-oriented conformations may only be induced when a low-affinity cation binding site is loaded. We propose that cations bound to high-affinity sites may be translocated together with cations bound to low-affinity sites or that they may block the translocation mechanism.


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Mutación , Animales , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/química , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Cationes , Unión Proteica , Ratas
13.
Mol Cell Biol ; 25(1): 78-87, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15601832

RESUMEN

The product of the intronless single copy gene RSC1A1, named RS1, is an intracellular 617-amino-acid protein that is involved in the regulation of the Na(+)-d-glucose cotransporter SGLT1. We generated and characterized RS1 knockout (RS1(-/-) mice. In the small intestines of RS1(-/-) mice, the SGLT1 protein was up-regulated sevenfold compared to that of wild-type mice but was not changed in the kidneys. The up-regulation of SGLT1 was posttranscriptional. Small intestinal d-glucose uptake measured in jointly perfused small bowel and liver was increased twofold compared to that of the wild-type, with increased peak concentrations of d-glucose in the portal vein. At birth, the weights of RS1(-/-) and wild-type mice were similar. At the age of 3 months, male RS1(-/-) mice had 5% higher weights and 15% higher food intakes, whereas their energy expenditures and serum leptin concentrations were similar to those of wild-type mice. At the age of 5 months, male and female RS1(-/-) mice were obese, with 30% increased body weight, 80% increased total fat, and 30% increased serum cholesterol. At this age, serum leptin was increased, whereas food intake was the same as for wild-type mice. The data suggest that the removal of RS1 leads to leptin-independent up-regulation of food intake, which causes obesity.


Asunto(s)
Glucosa/metabolismo , Intestino Delgado/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas de Transporte de Monosacáridos/fisiología , Obesidad/genética , Sodio/metabolismo , Regulación hacia Arriba , Animales , Transporte Biológico , Northern Blotting , Southern Blotting , Western Blotting , Colesterol/sangre , Clonación Molecular , Ensayo de Inmunoadsorción Enzimática , Femenino , Transportador de Glucosa de Tipo 2 , Insulina/metabolismo , Mucosa Intestinal/metabolismo , Intrones , Leptina/metabolismo , Masculino , Ratones , Ratones Noqueados , Microscopía Fluorescente , Modelos Genéticos , Fenotipo , Reacción en Cadena de la Polimerasa , Procesamiento Postranscripcional del ARN , Factores Sexuales , Transportador 1 de Sodio-Glucosa , Factores de Tiempo , Transcripción Genética , Transfección
14.
Pharmacogenetics ; 12(8): 591-5, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12439218

RESUMEN

By systematic mutation screening of the polyspecific organic cation transporter hOCT1 (SLC22A1) in 57 Caucasians, 25 genetic variations were identified and further analysed for population frequency. Five mutations resulting in the amino acid changes Arg61Cys, Cys88Arg, Phe160Leu, Gly401Ser, and Met420del, with respective allele frequencies of 9.1, 0.6, 22, 3.2, and 16%, were functionally characterized upon expression in Xenopus oocytes. Phe160Leu and Met420del exhibited substrate affinities and selectivites identical to hOCT1 wild-type. In contrast, uptake of 0.1 microm [3H]1-methyl-4-phenylpyridinium ([3H]MPP) by Arg61Cys, Cys88Arg and Gly401Ser were reduced to 30, 1.4 and 0.9% compared to wild-type, respectively. Since transport of 1 microm [3H]serotonin by Cys88Arg and Gly401Ser was reduced to only 13 and 12% of wild-type, these mutants exhibit a changed substrate selectivity. The data show that the mutants Arg61Cys, Cys88Arg and Gly401Ser could affect the disposition of OCT1 substrates and as a consequence may alter the duration and intensity of effects of drugs and neurotransmitters which are substrates for hOCT1.


Asunto(s)
Variación Genética , Transportador 1 de Catión Orgánico/fisiología , Adulto , Animales , Secuencia de Bases , Cartilla de ADN , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutagénesis Sitio-Dirigida , Mutación Missense , Transportador 1 de Catión Orgánico/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Xenopus
15.
Diabetes ; 61(1): 187-96, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22124465

RESUMEN

To clarify the physiological role of Na(+)-D-glucose cotransporter SGLT1 in small intestine and kidney, Sglt1(-/-) mice were generated and characterized phenotypically. After gavage of d-glucose, small intestinal glucose absorption across the brush-border membrane (BBM) via SGLT1 and GLUT2 were analyzed. Glucose-induced secretion of insulinotropic hormone (GIP) and glucagon-like peptide 1 (GLP-1) in wild-type and Sglt1(-/-) mice were compared. The impact of SGLT1 on renal glucose handling was investigated by micropuncture studies. It was observed that Sglt1(-/-) mice developed a glucose-galactose malabsorption syndrome but thrive normally when fed a glucose-galactose-free diet. In wild-type mice, passage of D-glucose across the intestinal BBM was predominantly mediated by SGLT1, independent the glucose load. High glucose concentrations increased the amounts of SGLT1 and GLUT2 in the BBM, and SGLT1 was required for upregulation of GLUT2. SGLT1 was located in luminal membranes of cells immunopositive for GIP and GLP-1, and Sglt1(-/-) mice exhibited reduced glucose-triggered GIP and GLP-1 levels. In the kidney, SGLT1 reabsorbed ∼3% of the filtered glucose under normoglycemic conditions. The data indicate that SGLT1 is 1) pivotal for intestinal mass absorption of d-glucose, 2) triggers the glucose-induced secretion of GIP and GLP-1, and 3) triggers the upregulation of GLUT2.


Asunto(s)
Glucosa/farmacocinética , Incretinas/metabolismo , Absorción Intestinal/genética , Transportador 1 de Sodio-Glucosa/fisiología , Animales , Femenino , Glucosa/farmacología , Glucosuria/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Intestino Delgado/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Transportador 1 de Sodio-Glucosa/genética , Transportador 1 de Sodio-Glucosa/metabolismo
16.
Am J Physiol Renal Physiol ; 296(4): F709-22, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19211691

RESUMEN

Uptake of substrate and electric charge was measured simultaneously in voltage-clamped Xenopus laevis oocytes expressing rat organic cation transporter 2 (rOCT2). At 0 mV, saturating substrate concentrations induced uptake of more positive elementary charges than monovalent organic cations, with charge-to-substrate ratios of 1.5 for guanidinium(+), 3.5 for tetraethylammonium(+), and 4.0 for 1-methyl-4-phenylpyridinium(+). At negative holding potentials, the charge-to-substrate ratios decreased toward unity. At 0 mV, charge-to-substrate ratios higher than unity were observed at different extracellular pH and after replacement of extracellular Na(+), K(+), Ca(2+), Mg(2+), and/or Cl(-). Charge-to-substrate ratios were not influenced by intracellular succinate(2-) or glutarate(2-). The effects of membrane potential and ion substitution strongly suggest that the surplus of transported positive charge is not generated by passive ion permeabilities. Rather, we hypothetize that small cations are taken up together with organic cation substrates whereas the outward reorientation of the empty transporter is electroneutral. Nonselective cotransport of small cations was supported by the three-dimensional structures of rOCT2 in its inward-facing and outward-facing conformations, which we determined by homology modeling based on known corresponding structures of H(+)-lactose permease of E. coli, and by functional analysis of OCT mutants. In our model, the innermost cavity of the outward-open binding cleft is negatively charged by Glu448 and Asp475, whereas the inward-open innermost cavity is electroneutral, containing Asp379, Asp475, Lys215, and Arg440. Substitution of Glu448 by glutamine reduced the charge-to-TEA(+) ratio at 0 mV to unity. The observed charge excess associated with organic cation uptake into depolarized cells may contribute to tubular damage in renal failure.


Asunto(s)
Proteínas de Transporte de Catión Orgánico/metabolismo , Animales , Sitios de Unión , Cationes , Ácidos Dicarboxílicos/metabolismo , Gluconatos/metabolismo , Ácido Glutámico , Glutamina , Concentración de Iones de Hidrógeno , Transporte Iónico , Ácido Láctico/metabolismo , Potenciales de la Membrana , Modelos Moleculares , Mutación , Oocitos , Proteínas de Transporte de Catión Orgánico/química , Proteínas de Transporte de Catión Orgánico/genética , Transportador 2 de Cátion Orgánico , Permeabilidad , Conformación Proteica , Ratas , Relación Estructura-Actividad , Tetraetilamonio/metabolismo , Xenopus laevis
17.
Biochemistry ; 47(15): 4552-64, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18361503

RESUMEN

Polyspecific organic cation and anion transporters of the SLC22 protein family are critically involved in absorption and excretion of drugs. To elucidate transport mechanisms, functional and biophysical characterization of purified transporters is required and tertiary structures must be determined. Here, we synthesized rat organic cation transporters OCT1 and OCT2 and rat organic anion transporter OAT1 in a cell free system in the absence of detergent. We solubilized the precipitates with 2% 1-myristoyl-2-hydroxy- sn-glycero-3-[phospho- rac-(1-glycerol)] (LMPG), purified the transporters in the presence of 1% 3-[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonate (CHAPS) or octyl glucoside, and reconstituted them into proteoliposomes. From 1 mL reaction vessels 0.13-0.36 mg of transporter proteins was purified. Thus, from five to ten 1 mL reaction vessels sufficient protein for crystallization was obtained. In the presence of 1% LMPG and 0.5% CHAPS, OCT1 and OAT1 formed homo-oligomers but no hetero-oligomers. After reconstitution of OCT1, OCT2, and OAT1 into proteoliposomes, similar Michaelis-Menten K m values were measured for uptake of 1-methyl-4-phenylpyridinium and p-aminohippurate (PAH (-)) by the organic cation and anion transporters, respectively, as after expression of the transporters in cells. Using the reconstituted system, evidence was obtained that OAT1 operates as obligatory and electroneutral PAH (-)/dicarboxylate antiporter and contains a low-affinity chloride binding site that stimulates turnover. PAH (-) uptake was observed only with alpha-ketoglutarate (KG (2-)) on the trans side, and trans-KG (2-) increased the PAH (-) concentration in voltage-clamped proteoliposomes transiently above equilibrium. The V max of PAH (-)/KG (2-) antiport was increased by Cl (-) in a manner independent of gradients, and PAH (-)/KG (2-) antiport was independent of membrane potential in the absence or presence of Cl (-).


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , 1-Metil-4-fenilpiridinio/metabolismo , Animales , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/aislamiento & purificación , Sistema Libre de Células , Ácidos Cetoglutáricos/metabolismo , Proteína 1 de Transporte de Anión Orgánico/genética , Proteína 1 de Transporte de Anión Orgánico/aislamiento & purificación , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/aislamiento & purificación , Transportador 2 de Cátion Orgánico , Proteolípidos/metabolismo , Ratas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Ácido p-Aminohipúrico/metabolismo
18.
Am J Physiol Renal Physiol ; 293(3): F767-79, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17567940

RESUMEN

Effects of the sulfhydryl reagent methylmethanethiosulfonate (MMTS) on functions of organic cation transporters (OCTs) were investigated. Currents induced by 10 mM choline [I(max(choline))] in Xenopus laevis oocytes expressing rat OCT1 (rOCT1) were increased four- to ninefold after 30-s incubation with 5 mM MMTS whereas I(max(choline)) by rat OCT2 was 70% decreased. MMTS activated the rOCT1 transporter within the plasma membrane without changing stoichiometry between translocated charge and cation. After modification of oocytes expressing rOCT1 or rOCT2 with MMTS, I(0.5(choline)) values for choline-induced currents were increased. For rOCT1 it was shown that MMTS increased I(0.5) values for different cations by different degrees. Mutagenesis of individual cysteine residues in rOCT1 revealed that modification of cysteine 322 in the large intracellular loop, and of cysteine 451 at the transition of the transmembrane alpha-helix (TMH) 10 to the short intracellular loop between the TMH 10 and 11 is responsible for the observed effects of MMTS. After replacement of cysteine 451 by methionine, the IC(50(choline)) for choline to inhibit MPP uptake by rOCT1 was increased whereas the I(0.5(choline)) value for choline-induced current remained unchanged. At variance, in double mutant Cys322Ser, Cys451Met, I(0.5(choline)) was increased compared with rOCT1 wild-type whereas in the single mutant Cys322Ser I(0.5(choline)) was not changed. The data suggest that modification of rOCT1 at cysteines 322 and 451 leads to an increase in turnover. They indicate that cysteine 451 in rOCT1 interacts with the large intracellular loop and that cysteine 451 in both rOCT1 and rOCT2 is critical for the affinity of choline.


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Cisteína/química , Proteínas de Transporte de Catión Orgánico/genética , Animales , Transporte Biológico Activo , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/química , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Capacidad Eléctrica , Electrofisiología , Exocitosis , Metilmetanosulfonato/farmacología , Modelos Moleculares , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Proteínas de Transporte de Catión Orgánico/química , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 2 de Cátion Orgánico , Unión Proteica , Conformación Proteica , Ratas , Especificidad por Sustrato , Xenopus laevis
19.
J Biol Chem ; 282(39): 28501-28513, 2007 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-17686765

RESUMEN

The human gene RSC1A1 codes for a 67-kDa protein named RS1 that mediates transcriptional and post-transcriptional regulation of Na(+)-D-glucose cotransporter SGLT1. The post-transcriptional regulation occurs at the trans-Golgi network (TGN). We identified two tripeptides in human RS1 (Gln-Cys-Pro (QCP) and Gln-Ser-Pro (QSP)) that induce posttranscriptional down-regulation of SGLT1 at the TGN leading to 40-50% reduction of SGLT1 in plasma membrane. For effective intracellular concentrations IC(50) values of 2.0 nM (QCP) and 0.16 nm (QSP) were estimated. Down-regulation of SGLT1 by tripeptides was attenuated by intracellular monosaccharides including non-metabolized methyl-alpha-D-glucopyranoside and 2-deoxyglucose. In small intestine post-transcriptional regulation of SGLT1 may contribute to glucose-dependent regulation of liver metabolism and intestinal mobility. QCP and QSP are transported by the H(+)-peptide cotransporter PepT1 that is colocated with SGLT1 in small intestinal enterocytes. Using coexpression of SGLT1 and PepT1 in Xenopus oocytes or polarized Caco-2 cells that contain both transporters we demonstrated that the tripeptides were effective when applied to the extracellular compartment. After a 1-h perfusion of intact rat small intestine with QSP, glucose absorption was reduced by 30%. The data indicate that orally applied tripeptides can be used to down-regulate small intestinal glucose absorption, e.g. in diabetes mellitus.


Asunto(s)
Glucosa/metabolismo , Absorción Intestinal/efectos de los fármacos , Proteínas de Transporte de Monosacáridos/metabolismo , Oligopéptidos/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transportador 1 de Sodio-Glucosa/metabolismo , Animales , Antimetabolitos/farmacología , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/fisiología , Células CACO-2 , Desoxiglucosa/farmacología , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Femenino , Motilidad Gastrointestinal/efectos de los fármacos , Motilidad Gastrointestinal/fisiología , Expresión Génica , Humanos , Absorción Intestinal/fisiología , Hígado/metabolismo , Masculino , Metilglucósidos/farmacología , Proteínas de Transporte de Monosacáridos/química , Proteínas de Transporte de Monosacáridos/genética , Oligopéptidos/química , Oligopéptidos/uso terapéutico , Oocitos/citología , Transportador de Péptidos 1 , Procesamiento Proteico-Postraduccional/fisiología , Ratas , Ratas Wistar , Transportador 1 de Sodio-Glucosa/genética , Simportadores/genética , Simportadores/metabolismo , Xenopus laevis , Red trans-Golgi/metabolismo
20.
Am J Physiol Renal Physiol ; 291(6): F1213-23, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16788146

RESUMEN

The product of gene RSC1A1, named RS1, participates in transcriptional and posttranscriptional regulation of the sodium-d-glucose cotransporter SGLT1. Using coexpression in oocytes of Xenopus laevis, posttranscriptional inhibition of human SGLT1 (hSGLT1) and some other transporters by human RS1 (hRS1) was demonstrated previously. In the present study, histidine-tagged hRS1 was expressed in oocytes or Sf9 cells and purified using nickel(II)-charged nitrilotriacetic acid-agarose. hRS1 protein was injected into oocytes expressing hSGLT1 or the human organic cation transporter hOCT2, and the effect on hSGLT1-mediated uptake of methyl-alpha-D-[14C]glucopyranoside ([14C]AMG) or hOCT2-mediated uptake of [14C]tetraethylammonium ([14C]TEA) was measured. Within 30 min after the injection of hRS1 protein, hSGLT1-expressed AMG uptake or hOCT2-expressed TEA uptake was inhibited by approximately 50%. Inhibition of AMG uptake was decreased when a dominant negative mutant of dynamin I was coexpressed and increased after stimulation of PKC. Inhibition remained unaltered when endocytosis was inhibited by chlorpromazine, imipramine, or filipin but was prevented when exocytosis was inhibited by botulinum toxin B or when the release of vesicles from the TGN and endosomes was inhibited by brefeldin A. Inhibition of hSGLT1-mediated AMG uptake and hOCT2-mediated TEA uptake by hRS1 protein were decreased at an enhanced intracellular AMG concentration. The data suggest that hRS1 protein exhibits glucose-dependent, short-term inhibition of hSGLT1 and hOCT2 by inhibiting the release of vesicles from the trans-Golgi network.


Asunto(s)
Exocitosis/fisiología , Proteínas de Transporte de Monosacáridos/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Brefeldino A/farmacología , Radioisótopos de Carbono , Dinaminas/metabolismo , Exocitosis/efectos de los fármacos , Glucosa/metabolismo , Humanos , Insectos , Metilglucósidos/farmacocinética , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/farmacología , Oocitos/citología , Oocitos/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 2 de Cátion Orgánico , Proteína Quinasa C/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Ratas , Sodio/metabolismo , Xenopus laevis , Red trans-Golgi/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA